1
|
Turner-Ivey B, Jenkins DP, Carroll SL. Multiple Roles for Neuregulins and Their ERBB Receptors in Neurodegenerative Disease Pathogenesis and Therapy. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00119-1. [PMID: 40254133 DOI: 10.1016/j.ajpath.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/06/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025]
Abstract
The role that neurotrophins, such as nerve growth factor, play in the pathogenesis of neurodegenerative diseases has long been appreciated. However, the neuregulin (NRG) family of growth factors and/or their v-erb-B2 avian erythroblastic leukemia viral oncogene homolog (ERBB) receptors have also been implicated in the pathogenesis of conditions, such as Alzheimer disease (AD), frontotemporal lobar degeneration (FTLD), and amyotrophic lateral sclerosis (ALS). In this review, we consider i) the structural variability of NRG isoforms generated by alternative RNA splicing, the use of multiple promoters and proteolysis, and the impact that this structural variability has on neuronal and glial physiology during development and adulthood. We discuss ii) the NRG receptors ERBB2, ERBB3, and ERBB4, how activation of each of these receptors further diversifies NRG actions in the central nervous system, and how dementia-related proteins, such as γ-secretase modulate the action of NRGs and their ERBB receptors. We then iii) turn to the abnormalities in NRG and ERBB expression and function evident in human AD and mouse AD models, how these abnormalities affect brain function, and attempts to use NRGs to treat AD. Finally, iv) we discuss NRG effects on the survival and function of neurons relevant to FTLD and ALS, alterations in NRG/ERBB signaling identified in these conditions, and the recent discovery of multiple human pedigrees in which autosomal dominant FTLD/ALS potentially results from point mutations in ERBB4.
Collapse
Affiliation(s)
- Brittany Turner-Ivey
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Dorea P Jenkins
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Steven L Carroll
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
2
|
Fisher ML, Prantzalos ER, O'Donovan B, Anderson TL, Sahoo PK, Twiss JL, Ortinski PI, Turner JR. Dynamic effects of ventral hippocampal NRG3/ERBB4 signaling on nicotine withdrawal-induced responses. Neuropharmacology 2024; 247:109846. [PMID: 38211698 PMCID: PMC10923109 DOI: 10.1016/j.neuropharm.2024.109846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/18/2023] [Accepted: 01/08/2024] [Indexed: 01/13/2024]
Abstract
Tobacco smoking remains a leading cause of preventable death in the United States, with approximately a 5% success rate for smokers attempting to quit. High relapse rates have been linked to several genetic factors, indicating that the mechanistic relationship between genes and drugs of abuse is a valuable avenue for the development of novel smoking cessation therapies. For example, various single nucleotide polymorphisms (SNPs) in the gene for neuregulin 3 (NRG3) and its cognate receptor, the receptor tyrosine-protein kinase erbB-4 (ERBB4), have been linked to nicotine addiction. Our lab has previously shown that ERBB4 plays a role in anxiety-like behavior during nicotine withdrawal (WD); however, the neuronal mechanisms and circuit-specific effects of NRG3-ERBB4 signaling during nicotine and WD are unknown. The present study utilizes genetic, biochemical, and functional approaches to examine the anxiety-related behavioral and functional role of NRG3-ERBB4 signaling, specifically in the ventral hippocampus (VH) of male and female mice. We report that 24hWD from nicotine is associated with altered synaptic expression of VH NRG3 and ERBB4, and genetic disruption of VH ErbB4 leads to an elimination of anxiety-like behaviors induced during 24hWD. Moreover, we observed attenuation of GABAergic transmission as well as alterations in Ca2+-dependent network activity in the ventral CA1 area of VH ErbB4 knock-down mice during 24hWD. Our findings further highlight contributions of the NRG3-ERBB4 signaling pathway to anxiety-related behaviors seen during nicotine WD.
Collapse
Affiliation(s)
- Miranda L Fisher
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, KY, USA
| | - Emily R Prantzalos
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, KY, USA
| | - Bernadette O'Donovan
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC, USA
| | - Tanner L Anderson
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Pabitra K Sahoo
- Department of Biological Sciences, University of South Carolina College of Arts and Sciences, Columbia, SC, USA
| | - Jeffery L Twiss
- Department of Biological Sciences, University of South Carolina College of Arts and Sciences, Columbia, SC, USA
| | - Pavel I Ortinski
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Jill R Turner
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, KY, USA.
| |
Collapse
|
3
|
Huang J, Zhang YY, Qiu YY, Yao S, Qiu WT, Peng JL, Li YQ, You QL, Wu CH, Wu EJ, Wang J, Zhou YL, Ning YP, Wang HS, Chen WB, Hu BJ, Liu Y, Sun XD. NRG1-ErbB4 signaling in the medial amygdala controls mating motivation in adult male mice. Cell Rep 2024; 43:113905. [PMID: 38446660 DOI: 10.1016/j.celrep.2024.113905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 12/12/2023] [Accepted: 02/19/2024] [Indexed: 03/08/2024] Open
Abstract
Motivation-driven mating is a basic affair for the maintenance of species. However, the underlying molecular mechanisms that control mating motivation are not fully understood. Here, we report that NRG1-ErbB4 signaling in the medial amygdala (MeA) is pivotal in regulating mating motivation. NRG1 expression in the MeA negatively correlates with the mating motivation levels in adult male mice. Local injection and knockdown of MeA NRG1 reduce and promote mating motivation, respectively. Consistently, knockdown of MeA ErbB4, a major receptor for NRG1, and genetic inactivation of its kinase both promote mating motivation. ErbB4 deletion decreases neuronal excitability, whereas chemogenetic manipulations of ErbB4-positive neuronal activities bidirectionally modulate mating motivation. We also identify that the effects of NRG1-ErbB4 signaling on neuronal excitability and mating motivation rely on hyperpolarization-activated cyclic nucleotide-gated channel 3. This study reveals a critical molecular mechanism for regulating mating motivation in adult male mice.
Collapse
Affiliation(s)
- Jie Huang
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Yan-Yan Zhang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, and Emergency Department of the Second Affiliated Hospital, School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Yu-Yang Qiu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, and Emergency Department of the Second Affiliated Hospital, School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Shan Yao
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, and Emergency Department of the Second Affiliated Hospital, School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Wan-Ting Qiu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, and Emergency Department of the Second Affiliated Hospital, School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Jin-Lin Peng
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yuan-Quan Li
- Department of Neurology, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Qiang-Long You
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, and Emergency Department of the Second Affiliated Hospital, School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Cui-Hong Wu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, and Emergency Department of the Second Affiliated Hospital, School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Er-Jian Wu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, and Emergency Department of the Second Affiliated Hospital, School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Jin Wang
- Department of Physiology, Guangxi University of Science and Technology, Liuzhou, China
| | - Yan-Ling Zhou
- Department of Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yu-Ping Ning
- Department of Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hong-Sheng Wang
- Songjiang Research Institute, Shanghai Songjiang District Central Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wen-Bing Chen
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, and Emergency Department of the Second Affiliated Hospital, School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Bing-Jie Hu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, and Emergency Department of the Second Affiliated Hospital, School of Basic Medicine, Guangzhou Medical University, Guangzhou, China.
| | - Youtan Liu
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China.
| | - Xiang-Dong Sun
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
4
|
Fisher ML, Prantzalos ER, O'Donovan B, Anderson T, Sahoo PK, Twiss JL, Ortinski PI, Turner JR. Dynamic Effects of Ventral Hippocampal NRG3/ERBB4 Signaling on Nicotine Withdrawal-Induced Responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.17.524432. [PMID: 36711798 PMCID: PMC9882308 DOI: 10.1101/2023.01.17.524432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Tobacco smoking remains a leading cause of preventable death in the United States, with a less than 5% success rate for smokers attempting to quit. High relapse rates have been linked to several genetic factors, indicating that the mechanistic relationship between genes and drugs of abuse is a valuable avenue for the development of novel smoking cessation therapies. For example, various single nucleotide polymorphisms (SNPs) in the gene for neuregulin 3 (NRG3) and its cognate receptor, the receptor tyrosine-protein kinase erbB-4 (ERBB4), have been linked to nicotine addiction. Our lab has previously shown that ERBB4 plays a role in anxiety-like behavior during nicotine withdrawal (WD); however, the neuronal mechanisms and circuit-specific effects of NRG3-ERBB4 signaling during nicotine and WD are unknown. The present study utilizes genetic, biochemical, and functional approaches to examine the anxiety-related behavioral and functional role of NRG3-ERBB4 signaling, specifically in the ventral hippocampus (VH). We report that 24hWD from nicotine is associated with altered synaptic expression of VH NRG3 and ERBB4, and genetic disruption of VH ErbB4 leads to an elimination of anxiety-like behaviors induced during 24hWD. Moreover, we observed attenuation of GABAergic transmission as well as alterations in Ca2+-dependent network activity in the ventral CA1 area of VH ErbB4 knock-down mice during 24hWD. Our findings further highlight contributions of the NRG3-ERBB4 signaling pathway to anxiety-related behaviors seen during nicotine WD.
Collapse
Affiliation(s)
- Miranda L Fisher
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| | - Emily R Prantzalos
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| | - Bernadette O'Donovan
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Tanner Anderson
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Pabitra K Sahoo
- Department of Biological Sciences, University of South Carolina College of Arts and Sciences, Columbia, South Carolina, USA
| | - Jeffery L Twiss
- Department of Biological Sciences, University of South Carolina College of Arts and Sciences, Columbia, South Carolina, USA
| | - Pavel I Ortinski
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Jill R Turner
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| |
Collapse
|
5
|
Chan CK, Lim KS, Low SK, Tan CT, Ng CC. Genetic interaction between GABRA1 and ERBB4 variants in the pathogenesis of genetic generalized epilepsy. Epilepsy Res 2023; 189:107070. [PMID: 36584483 DOI: 10.1016/j.eplepsyres.2022.107070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/06/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022]
Abstract
Epilepsy is a complex neurological disease that can be caused by both genetic and environmental factors. Many studies have been conducted to investigate the genetic risk variants and molecular mechanisms of epilepsy. Disruption of excitation-inhibition balance (E/I balance) is one of the widely accepted disease mechanisms of epilepsy. The maintenance of E/I balance is an intricate process that is governed by multiple proteins. Using whole exome sequencing (WES), we identified a novel GABRA1 c.448G>A (p.E150K) variant and ERBB4 c.1972A>T (p.I658F, rs190654033) variant in a Malaysian Chinese family with genetic generalized epilepsy (GGE). The GGE may be triggered by dysregulation of E/I balance mechanism. Segregation of the variants in the family was verified by Sanger sequencing. All family members with GGE inherited both variants. However, family members who carried only one of the variants did not show any symptoms of GGE. Both the GABRA1 and ERBB4 variants were predicted damaging by MutationTaster and CADD, and protein structure analysis showed that the variants had resulted in the formation of additional hydrogen bonds in the mutant proteins. GABRA1 variant could reduce the efficiency of GABAA receptors, and constitutively active ERBB4 receptors caused by the ERBB4 variant promote internalization of GABAA receptors. The interaction between the two variants may cause a greater disruption in E/I balance, which is more likely to induce a seizure. Nevertheless, this disease model was derived from a single small family, further studies are still needed to confirm the verifiability of the purported disease model.
Collapse
Affiliation(s)
- Chung-Kin Chan
- Genetics and Molecular Biology, Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur, Malaysia; Division of Neurology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Kheng-Seang Lim
- Division of Neurology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia.
| | - Siew-Kee Low
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Chong-Tin Tan
- Division of Neurology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Ching-Ching Ng
- Genetics and Molecular Biology, Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
6
|
Pitcher JL, Alexander N, Miranda PJ, Johns TG. ErbB4 in the brain: Focus on high grade glioma. Front Oncol 2022; 12:983514. [PMID: 36119496 PMCID: PMC9471956 DOI: 10.3389/fonc.2022.983514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) family of receptor tyrosine kinases (RTKs) consists of EGFR, ErbB2, ErbB3, and ErbB4. These receptors play key roles in cell proliferation, angiogenesis, cell migration, and in some cases, tumor promotion. ErbB4 is a unique member of the EGFR family, implicated not only in pro-tumorigenic mechanisms, such as cell proliferation and migration, but also in anti-tumorigenic activities, including cell differentiation and apoptosis. ErbB4 is differentially expressed in a wide variety of tissues, and interestingly, as different isoforms that result in vastly different signalling outcomes. Most studies have either ignored the presence of these isoforms or used overexpression models that may mask the true function of ErbB4. ErbB4 is widely expressed throughout the body with significant expression in skeletal tissue, mammary glands, heart, and brain. Knockout models have demonstrated embryonic lethality due to disrupted heart and brain development. Despite high expression in the brain and a critical role in brain development, remarkably little is known about the potential signalling activity of ErbB4 in brain cancer.This review focuses on the unique biology of ErbB4 in the brain, and in particular, highlights brain cancer research findings. We end the review with a focus on high grade gliomas, primarily glioblastoma, a disease that has been shown to involve EGFR and its mutant forms. The role of the different ErbB4 isotypes in high grade gliomas is still unclear and future research will hopefully shed some light on this question.
Collapse
Affiliation(s)
- Jamie-Lee Pitcher
- Oncogenic Signalling Laboratory, Telethon Kids Institute, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
- *Correspondence: Jamie-Lee Pitcher,
| | - Naomi Alexander
- Oncogenic Signalling Laboratory, Telethon Kids Institute, Nedlands, WA, Australia
| | - Panimaya Jeffreena Miranda
- Oncogenic Signalling Laboratory, Telethon Kids Institute, Nedlands, WA, Australia
- Division of Paediatrics/Centre for Child Health Research, University of Western Australia, Crawley, WA, Australia
| | - Terrance G. Johns
- Oncogenic Signalling Laboratory, Telethon Kids Institute, Nedlands, WA, Australia
- Division of Paediatrics/Centre for Child Health Research, University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
7
|
NKCC1 to KCC2 mRNA Ratio in Schizophrenia and Its Psychopathology: a Case-Control Study. J Mol Neurosci 2022; 72:1670-1681. [PMID: 35624355 DOI: 10.1007/s12031-021-01879-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 06/21/2021] [Indexed: 10/18/2022]
Abstract
Schizophrenia (SCZ) is a debilitating, destructive, and chronic mental disorder and affects approximately one percent of the human population. Diagnosis in psychiatry is based on the patient's descriptions of his/her symptoms, interviewer's observations, history of disorder over time, and response to treatment. All of these data measure phenotype-based functions. But it appears that accurate diagnosis of such a complex disorder must be based on valid and reliable factors. In the present study, gene selection was based on the possible role of γ-aminobutyric acid (GABA) in psychopathology of SCZ and expression in blood. We evaluated the association of Na+-K+-Cl- co-transporter 1 (NKCC1) and K+-Cl- co-transporter 2 (KCC2) genes' messenger ribonucleic acid (mRNA) levels, and also the NKCC1/KCC2 ratio with positive and negative syndrome scale (PANSS) and brief psychiatric rating scale (BPRS) scores in an SCZ group. By using real-time PCR (RT-PCR), the present study is the first attempt to explore levels of NKCC1 and KCC2 expression at mRNA level and their relative expression in human peripheral blood of patients with SCZ. Our results showed that the NKCC1 to KCC2 mRNA ratio is significantly increased (but based on the delta cycle of threshold [∆Ct] is significantly lower) in the total sample of cases rather than controls (p = 0.045) and also higher in male sample cases rather than male controls (p = 0.016). In female samples, we found a trend toward a significant effect between the case and control participants (p = 0.075). We also found statistically significant association between mRNA of NKCC1 and KCC2 genes and NKCC1/KCC2 mRNA ratio with the positive and negative syndrome scale (PANSS) and brief psychiatric rating scale (BPRS) scores.
Collapse
|
8
|
Erben L, Welday JP, Cronin ME, Murphy R, Skirzewski M, Vullhorst D, Carroll SL, Buonanno A. Developmental, neurochemical, and behavioral analyses of ErbB4 Cyt-1 knockout mice. J Neurochem 2022; 161:435-452. [PMID: 35523590 PMCID: PMC9149141 DOI: 10.1111/jnc.15612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 01/26/2023]
Abstract
Neuregulins (NRGs) and their cognate neuronal receptor ERBB4, which is expressed in GABAergic and dopaminergic neurons, regulate numerous behaviors in rodents and have been identified as schizophrenia at-risk genes. ErbB4 transcripts are alternatively spliced to generate isoforms that either include (Cyt-1) or exclude (Cyt-2) exon 26, which encodes a cytoplasmic domain that imparts ErbB4 receptors the ability to signal via the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway. Although ErbB4 Cyt-1/2 isoforms have been studied in transfected cultured cells, their functions in vivo remain unknown. Here, we generated ErbB4-floxed (ErbB4-Cyt1fl/fl ) mice to investigate the effects of germline (constitutive) and conditional (acute) deletions of the Cyt-1 exon. Overall receptor mRNA levels remain unchanged in germline ErbB4 Cyt-1 knockouts (Cyt-1 KOs), with all transcripts encoding Cyt-2 variants. In contrast to mice lacking all ErbB4 receptor function, GABAergic interneuron migration and number are unaltered in Cyt-1 KOs. However, basal extracellular dopamine (DA) levels in the medial prefrontal cortex are increased in Cyt-1 heterozygotes. Despite these neurochemical changes, Cyt-1 heterozygous and homozygous mice do not manifest behavioral abnormalities previously reported to be altered in ErbB4 null mice. To address the possibility that Cyt-2 variants compensate for the lack of Cyt-1 during development, we microinjected an adeno-associated virus expressing Cre-recombinase (AAV-Cre) into the DA-rich ventral tegmental area of adult ErbB4-Cyt1fl/fl mice to acutely target exon 26. These conditional Cyt-1 KOs were found to exhibit behavioral abnormalities in the elevated plus maze and startle response, consistent with the idea that late exon 26 ablations may circumvent compensation by Cyt-2 variants. Taken together, our observations indicate that ErbB4 Cyt-1 function in vivo is important for DA balance and behaviors in adults.
Collapse
Affiliation(s)
- Larissa Erben
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, Maryland, USA
| | - Jacqueline P Welday
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, Maryland, USA
| | - Marie E Cronin
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, Maryland, USA
| | - Ricardo Murphy
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, Maryland, USA
| | - Miguel Skirzewski
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, Maryland, USA
| | - Detlef Vullhorst
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, Maryland, USA
| | - Steven L Carroll
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Andres Buonanno
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, Maryland, USA
| |
Collapse
|
9
|
Belonwu SA, Li Y, Bunis DG, Rao AA, Solsberg CW, Oskotsky T, Taubes AL, Grone B, Zalocusky KA, Fragiadakis GK, Huang Y, Sirota M. Bioinformatics Analysis of Publicly Available Single-Nuclei Transcriptomics Alzheimer’s Disease Datasets Reveals APOE Genotype-Specific Changes Across Cell Types in Two Brain Regions. Front Aging Neurosci 2022; 14:749991. [PMID: 35572130 PMCID: PMC9093608 DOI: 10.3389/fnagi.2022.749991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s Disease (AD) is a complex neurodegenerative disease that gravely affects patients and imposes an immense burden on caregivers. Apolipoprotein E4 (APOE4) has been identified as the most common genetic risk factor for AD, yet the molecular mechanisms connecting APOE4 to AD are not well understood. Past transcriptomic analyses in AD have revealed APOE genotype-specific transcriptomic differences; however, these differences have not been explored at a single-cell level. To elucidate more complex APOE genotype-specific disease-relevant changes masked by the bulk analysis, we leverage the first two single-nucleus RNA sequencing AD datasets from human brain samples, including nearly 55,000 cells from the prefrontal and entorhinal cortices. In each brain region, we performed a case versus control APOE genotype-stratified differential gene expression analysis and pathway network enrichment in astrocytes, microglia, neurons, oligodendrocytes, and oligodendrocyte progenitor cells. We observed more global transcriptomic changes in APOE4 positive AD cells and identified differences across APOE genotypes primarily in glial cell types. Our findings highlight the differential transcriptomic perturbations of APOE isoforms at a single-cell level in AD pathogenesis and have implications for precision medicine development in the diagnosis and treatment of AD.
Collapse
Affiliation(s)
- Stella A. Belonwu
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
- Pharmaceutical Sciences and Pharmacogenomics Graduate Program, University of California, San Francisco, San Francisco, CA, United States
| | - Yaqiao Li
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
- Pharmaceutical Sciences and Pharmacogenomics Graduate Program, University of California, San Francisco, San Francisco, CA, United States
| | - Daniel G. Bunis
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
- CoLabs, University of California, San Francisco, San Francisco, CA, United States
- Bakar ImmunoX Initiative, University of California, San Francisco, San Francisco, CA, United States
| | - Arjun Arkal Rao
- CoLabs, University of California, San Francisco, San Francisco, CA, United States
- Bakar ImmunoX Initiative, University of California, San Francisco, San Francisco, CA, United States
- Department of Pathology, University of California, San Francisco, San Francisco, CA, United States
| | - Caroline Warly Solsberg
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
- Pharmaceutical Sciences and Pharmacogenomics Graduate Program, University of California, San Francisco, San Francisco, CA, United States
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
| | - Tomiko Oskotsky
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Alice L. Taubes
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
- Gladstone Institute of Neurological Disease, San Francisco, CA, United States
| | - Brian Grone
- Gladstone Institute of Neurological Disease, San Francisco, CA, United States
| | - Kelly A. Zalocusky
- Gladstone Institute of Neurological Disease, San Francisco, CA, United States
| | - Gabriela K. Fragiadakis
- CoLabs, University of California, San Francisco, San Francisco, CA, United States
- Bakar ImmunoX Initiative, University of California, San Francisco, San Francisco, CA, United States
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Yadong Huang
- Department of Pathology, University of California, San Francisco, San Francisco, CA, United States
- Gladstone Institute of Neurological Disease, San Francisco, CA, United States
- Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
- *Correspondence: Marina Sirota,
| |
Collapse
|
10
|
Asede D, Okoh J, Ali S, Doddapaneni D, Bolton MM. Deletion of ErbB4 Disrupts Synaptic Transmission and Long-Term Potentiation of Thalamic Input to Amygdalar Medial Paracapsular Intercalated Cells. Front Synaptic Neurosci 2021; 13:697110. [PMID: 34393751 PMCID: PMC8355744 DOI: 10.3389/fnsyn.2021.697110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 07/08/2021] [Indexed: 11/20/2022] Open
Abstract
Identification of candidate risk genes and alteration in the expression of proteins involved in regulating inhibitory neuron function in various psychiatric disorders, support the notion that GABAergic neuron dysfunction plays an important role in disease etiology. Genetic variations in neuregulin and its receptor kinase ErbB4, expressed exclusively by GABAergic neurons in the CNS, have been linked with schizophrenia. In the amygdala, ErbB4 is highly expressed in GABAergic intercalated cell clusters (ITCs), which play a critical role in amygdala-dependent behaviors. It is however unknown whether ErbB4 deletion from ITCs affects their synaptic properties and function in amygdala circuitry. Here, we examined the impact of ErbB4 deletion on inhibitory and excitatory circuits recruiting medial paracapsular ITCs (mpITCs) using electrophysiological techniques. Ablation of ErbB4 in mpITCs suppressed NMDA receptor-mediated synaptic transmission at thalamo-mpITC synapses and enhanced thalamic driven GABAergic transmission onto mpITCs. Furthermore, long-term potentiation (LTP) at thalamo-mpITC synapses was compromised in ErbB4 mutant mice, indicating that ErbB4 activity is critical for LTP at these synapses. Together, our findings suggest that ErbB4 deletion from mpITCs disrupts excitation-inhibition balance and learning mechanisms in amygdala circuits.
Collapse
Affiliation(s)
- Douglas Asede
- Disorders of Neural Circuit Function, Max Planck Florida Institute for Neuroscience, Jupiter, FL, United States
| | - James Okoh
- Disorders of Neural Circuit Function, Max Planck Florida Institute for Neuroscience, Jupiter, FL, United States
| | - Sabah Ali
- Disorders of Neural Circuit Function, Max Planck Florida Institute for Neuroscience, Jupiter, FL, United States
| | - Divyesh Doddapaneni
- Disorders of Neural Circuit Function, Max Planck Florida Institute for Neuroscience, Jupiter, FL, United States
| | - M McLean Bolton
- Disorders of Neural Circuit Function, Max Planck Florida Institute for Neuroscience, Jupiter, FL, United States
| |
Collapse
|
11
|
Luo B, Liu Z, Lin D, Chen W, Ren D, Yu Z, Xiong M, Zhao C, Fei E, Li B. ErbB4 promotes inhibitory synapse formation by cell adhesion, independent of its kinase activity. Transl Psychiatry 2021; 11:361. [PMID: 34226493 PMCID: PMC8257755 DOI: 10.1038/s41398-021-01485-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 06/04/2021] [Accepted: 06/10/2021] [Indexed: 12/12/2022] Open
Abstract
The precise control of the nervous system function under the vitality of synapses is extremely critical. Efforts have been taken to explore the underlying cellular and molecular mechanisms for synapse formation. Cell adhesion molecules have been found important for synapse assembly in the brain. Many trans-adhesion complexes have been identified to modulate excitatory synapse formation. However, little is known about the synaptogenic mechanisms for inhibitory synapses. ErbB4 is a receptor tyrosine kinase enriched in interneurons. Here, we showed that overexpressing ErbB4 in HEK293T cells induced gephyrin or GABAAR α1 puncta in co-cultured primary hippocampal neurons. This induction of ErbB4 was independent of its kinase activity. K751M, a kinase-dead mutant of ErbB4, can also induce gephyrin or GABAAR α1 puncta in the co-culture system. We further constructed K751M knock-in mice and found that the homozygous were viable at birth and fertile without changes in gross brain structure. The number of interneurons and inhibitory synapses onto pyramidal neurons (PyNs) were comparable between K751M and wild-type mice but decreased in ErbB4-Null mice. Moreover, ErbB4 can interact in trans with Slitrk3, a transmembrane postsynaptic protein at inhibitory synapses, through the extracellular RLD domain of ErbB4. The deletion of RLD diminished the induction of gephyrin or GABAAR α1 puncta by ErbB4. Finally, disruption of ErbB4-Slitrk3 interaction through neutralization of Slitrk3 by secretable RLD decreased inhibitory synapses onto PyNs and impaired GABAergic transmission. These results identify that ErbB4, as a cell adhesion molecule, promotes inhibitory synapse formation onto PyNs by interacting with Slitrk3 and in a kinase-independent manner, providing an unexpected mechanism of ErbB4 in inhibitory synapse formation.
Collapse
Affiliation(s)
- Bin Luo
- grid.260463.50000 0001 2182 8825School of Life Sciences, Nanchang University, Nanchang, China ,grid.260463.50000 0001 2182 8825Institute of Life Science, Nanchang University, Nanchang, China
| | - Ziyang Liu
- grid.260463.50000 0001 2182 8825School of Life Sciences, Nanchang University, Nanchang, China ,grid.260463.50000 0001 2182 8825Institute of Life Science, Nanchang University, Nanchang, China
| | - Dong Lin
- grid.260463.50000 0001 2182 8825School of Life Sciences, Nanchang University, Nanchang, China ,grid.260463.50000 0001 2182 8825Institute of Life Science, Nanchang University, Nanchang, China
| | - Wenbing Chen
- grid.260463.50000 0001 2182 8825School of Life Sciences, Nanchang University, Nanchang, China ,grid.260463.50000 0001 2182 8825Institute of Life Science, Nanchang University, Nanchang, China
| | - Dongyan Ren
- grid.260463.50000 0001 2182 8825School of Life Sciences, Nanchang University, Nanchang, China ,grid.260463.50000 0001 2182 8825Institute of Life Science, Nanchang University, Nanchang, China
| | - Zheng Yu
- grid.260463.50000 0001 2182 8825Institute of Life Science, Nanchang University, Nanchang, China
| | - Mingtao Xiong
- grid.260463.50000 0001 2182 8825School of Life Sciences, Nanchang University, Nanchang, China ,grid.260463.50000 0001 2182 8825Institute of Life Science, Nanchang University, Nanchang, China
| | - Changqin Zhao
- grid.260463.50000 0001 2182 8825School of Life Sciences, Nanchang University, Nanchang, China ,grid.260463.50000 0001 2182 8825Institute of Life Science, Nanchang University, Nanchang, China
| | - Erkang Fei
- School of Life Sciences, Nanchang University, Nanchang, China. .,Institute of Life Science, Nanchang University, Nanchang, China.
| | - Baoming Li
- School of Life Sciences, Nanchang University, Nanchang, China. .,Institute of Life Science, Nanchang University, Nanchang, China. .,Department of Psychology and Institute of Brain Science, School of Education, Hangzhou Normal University, Hangzhou, China.
| |
Collapse
|
12
|
Wang J, Huang J, Yao S, Wu JH, Li HB, Gao F, Wang Y, Huang GB, You QL, Li J, Chen X, Sun XD. The ketogenic diet increases Neuregulin 1 expression via elevating histone acetylation and its anti-seizure effect requires ErbB4 kinase activity. Cell Biosci 2021; 11:93. [PMID: 34020711 PMCID: PMC8139023 DOI: 10.1186/s13578-021-00611-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 05/11/2021] [Indexed: 12/16/2022] Open
Abstract
Background The ketogenic diet (KD)has been considered an effective treatment for epilepsy, whereas its underlying mechanisms remain obscure. We have previously reported that the KD feeding increased Neuregulin 1 (NRG1) expression in the hippocampus; disruption of NRG1 signaling by genetically deleting its receptor-ErbB4 abolished KDs effects on inhibitory synaptic activity and seizures. However, it is still unclear about the mechanisms underlying the effect of KD on NRG1 expression and whether the effects of KD require ErbB4 kinase activity. Methods The effects of the KD on NRG1 expression were assessed via western blotting and real-time PCR. Acetylation level at the Nrg1 promoter locus was examined using the chromatin immunoprecipitation technique. Kainic acid (KA)-induced acute seizure model was utilized to examine the effects of KD and histone deacetylase inhibitor-TSA on seizures. Synaptic activities in the hippocampus were recorded with the technique of electrophysiology. The obligatory role of ErbB4 kinase activity in KDs effects on seizures and inhibitory synaptic activity was evaluated by using ErbB kinase antagonist and transgenic mouse-T796G. Results We report that KD specifically increases Type I NRG1 expression in the hippocampus. Using the chromatin immunoprecipitation technique, we observe increased acetylated-histone occupancy at the Nrg1 promoter locus of KD-fed mice. Treatment of TSA dramatically elevates NRG1 expression and diminishes the difference between the effects of the control diet (CD) and KD. These data indicate that KD increases NRG1 expression via up-regulating histone acetylation. Moreover, both pharmacological and genetic inhibitions of ErbB4 kinase activity significantly block the KDs effects on inhibitory synaptic activity and seizure, suggesting an essential role of ErbB4 kinase activity. Conclusion These results strengthen our understanding of the role of NRG1/ErbB4 signaling in KD and shed light on novel therapeutic interventions for epilepsy. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00611-7.
Collapse
Affiliation(s)
- Jin Wang
- Emergency Department, Institute of Neuroscience, Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Jie Huang
- Emergency Department, Institute of Neuroscience, Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Shan Yao
- Emergency Department, Institute of Neuroscience, Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Jia-Hui Wu
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Hui-Bin Li
- Department of Pathology, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Feng Gao
- Emergency Department, Institute of Neuroscience, Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Ying Wang
- Emergency Department, Institute of Neuroscience, Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Guo-Bin Huang
- Emergency Department, Institute of Neuroscience, Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Qiang-Long You
- Emergency Department, Institute of Neuroscience, Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Jianhua Li
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Science, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Xiaohui Chen
- Emergency Department, Institute of Neuroscience, Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China.
| | - Xiang-Dong Sun
- Emergency Department, Institute of Neuroscience, Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China.
| |
Collapse
|
13
|
Hyder Z, Van Paesschen W, Sabir A, Sansbury FH, Burke KB, Khan N, Chandler KE, Cooper NS, Wright R, McHale E, Van Esch H, Banka S. ERBB4 exonic deletions on chromosome 2q34 in patients with intellectual disability or epilepsy. Eur J Hum Genet 2021; 29:1377-1383. [PMID: 33603162 PMCID: PMC8440581 DOI: 10.1038/s41431-021-00815-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 12/09/2020] [Accepted: 01/19/2021] [Indexed: 11/09/2022] Open
Abstract
ERBB4 encodes the tyrosine kinase receptor HER4, a critical regulator of normal cell function and neurodevelopmental processes in the brain. One of the key ligands of HER4 is neureglin-1 (NRG1), and the HER4-NRG1 signalling pathway is essential in neural crest cell migration, and neuronal differentiation. Pharmacological inactivation of HER4 has been shown to hasten the progression of epileptogenesis in rodent models, and heterozygous ERBB4 null mice are shown to have cognitive deficits and delayed motor development. Thus far there is only a single case report in the literature of a heterozygous ERBB4 deletion in a patient with intellectual disability (ID). We identified nine subjects from five unrelated families with chromosome 2q34 deletions, resulting in heterozygous intragenic loss of multiple exons of ERBB4, associated with either non-syndromic ID or generalised epilepsy. In one family, the deletion segregated with ID in five affected relatives. Overall, this case series further supports that haploinsufficiency of ERBB4 leads to non-syndromic intellectual disability or epilepsy.
Collapse
Affiliation(s)
- Zerin Hyder
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK.
| | - Wim Van Paesschen
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium.,Laboratory for Epilepsy Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Ataf Sabir
- West Midlands Regional Genetics Service, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Francis H Sansbury
- All Wales Medical Genomics Service, NHS Wales Cardiff and Vale University Health Board, Institute of Medical Genetics, University Hospital of Wales, Cardiff, UK
| | - Katherine B Burke
- All Wales Medical Genomics Service, NHS Wales Cardiff and Vale University Health Board, Institute of Medical Genetics, University Hospital of Wales, Cardiff, UK
| | - Naz Khan
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Kate E Chandler
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK.,Division of Evolution & Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Nicola S Cooper
- West Midlands Regional Genetics Service, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Ronnie Wright
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Edward McHale
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Hilde Van Esch
- Center for Human Genetics, University Hospitals Leuven, University of Leuven, Leuven, Belgium
| | - Siddharth Banka
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK.,Division of Evolution & Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
14
|
ErbB4 Null Mice Display Altered Mesocorticolimbic and Nigrostriatal Dopamine Levels as well as Deficits in Cognitive and Motivational Behaviors. eNeuro 2020; 7:ENEURO.0395-19.2020. [PMID: 32354758 PMCID: PMC7242816 DOI: 10.1523/eneuro.0395-19.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 12/17/2022] Open
Abstract
Natural genetic variants of Neuregulin1 (NRG1) and its cognate receptor ErbB4 are associated with a risk for schizophrenia. Whereas most studies on NRG1-ErbB4 signaling have focused on GABAergic interneurons, ErbB4 is also expressed by midbrain dopaminergic neurons where it modulates extracellular dopamine (DA) levels. Here, we report that extracellular steady-state levels of DA are reduced in the medial prefrontal cortex (mPFC; −65%), hippocampus (−53%) and nucleus accumbens (NAc; −35%), but are elevated in the dorsal striatum (+25%) of ErbB4 knock-out mice (ErbB4 KOs) relative to wild-type controls. This pattern of DA imbalance recapitulates the reported prefrontal cortical reduction and striatal increase of DA levels in schizophrenia patients. Next, we report on a battery of behavioral tasks used to evaluate locomotor, cognitive and motivational behaviors in ErbB4 KOs relative to controls. We found that ErbB4 KOs are hyperactive in a novel open field but not in their familiar home cage, are more sensitive to amphetamine, perform poorly in the T-maze and novel object recognition (NOR) tasks, exhibit reduced spatial learning and memory on the Barnes maze, and perform markedly worse in conditioned place preference (CPP) tasks when associating cued-reward palatable food with location. However, we found that the poor performance of ErbB4 KOs in CPP are likely due to deficits in spatial memory, instead of reward seeking, as ErbB4 KOs are more motivated to work for palatable food rewards. Our findings indicate that ErbB4 signaling affects tonic DA levels and modulates a wide array of behavioral deficits relevant to psychiatric disorders, including schizophrenia.
Collapse
|
15
|
Rahman A, Weber J, Labin E, Lai C, Prieto AL. Developmental expression of Neuregulin‐3 in the rat central nervous system. J Comp Neurol 2018; 527:797-817. [DOI: 10.1002/cne.24559] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/24/2018] [Accepted: 10/11/2018] [Indexed: 12/28/2022]
Affiliation(s)
- Afrida Rahman
- Departmentof Psychological and Brain SciencesIndiana University Bloomington Indiana
| | - Janet Weber
- Department NeuroscienceUniversity of California San Diego San Diego California
| | - Edward Labin
- Department of NeurologyUniversity of Minnesota Minneapolis
| | - Cary Lai
- Departmentof Psychological and Brain SciencesIndiana University Bloomington Indiana
| | - Anne L Prieto
- Departmentof Psychological and Brain SciencesIndiana University Bloomington Indiana
| |
Collapse
|
16
|
Müller T, Braud S, Jüttner R, Voigt BC, Paulick K, Sheean ME, Klisch C, Gueneykaya D, Rathjen FG, Geiger JR, Poulet JF, Birchmeier C. Neuregulin 3 promotes excitatory synapse formation on hippocampal interneurons. EMBO J 2018; 37:embj.201798858. [PMID: 30049711 PMCID: PMC6120667 DOI: 10.15252/embj.201798858] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 06/19/2018] [Accepted: 06/21/2018] [Indexed: 01/09/2023] Open
Abstract
Hippocampal GABAergic interneurons are crucial for cortical network function and have been implicated in psychiatric disorders. We show here that Neuregulin 3 (Nrg3), a relatively little investigated low-affinity ligand, is a functionally dominant interaction partner of ErbB4 in parvalbumin-positive (PV) interneurons. Nrg3 and ErbB4 are located pre- and postsynaptically, respectively, in excitatory synapses on PV interneurons in vivo Additionally, we show that ablation of Nrg3 results in a similar phenotype as the one described for ErbB4 ablation, including reduced excitatory synapse numbers on PV interneurons, altered short-term plasticity, and disinhibition of the hippocampal network. In culture, presynaptic Nrg3 increases excitatory synapse numbers on ErbB4+ interneurons and affects short-term plasticity. Nrg3 mutant neurons are poor donors of presynaptic terminals in the presence of competing neurons that produce recombinant Nrg3, and this bias requires postsynaptic ErbB4 but not ErbB4 kinase activity. Furthermore, when presented by non-neuronal cells, Nrg3 induces postsynaptic membrane specialization. Our data indicate that Nrg3 provides adhesive cues that facilitate excitatory neurons to synapse onto ErbB4+ interneurons.
Collapse
Affiliation(s)
- Thomas Müller
- Developmental Biology/Signal Transduction Group, Max-Delbrueck-Centrum in the Helmholtz Association, Berlin, Germany
| | - Stephanie Braud
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - René Jüttner
- Developmental Neurobiology Group, Max-Delbrueck-Centrum in the Helmholtz Association, Berlin, Germany
| | - Birgit C Voigt
- Neural Circuits and Behaviour Group, Max-Delbrueck-Centrum in the Helmholtz Association, Berlin, Germany
| | - Katharina Paulick
- Developmental Biology/Signal Transduction Group, Max-Delbrueck-Centrum in the Helmholtz Association, Berlin, Germany
| | - Maria E Sheean
- Developmental Biology/Signal Transduction Group, Max-Delbrueck-Centrum in the Helmholtz Association, Berlin, Germany
| | - Constantin Klisch
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dilansu Gueneykaya
- Cellular Neuroscience Group, Max-Delbrueck-Centrum in the Helmholtz Association, Berlin, Germany
| | - Fritz G Rathjen
- Developmental Neurobiology Group, Max-Delbrueck-Centrum in the Helmholtz Association, Berlin, Germany
| | - Jörg Rp Geiger
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - James Fa Poulet
- Neural Circuits and Behaviour Group, Max-Delbrueck-Centrum in the Helmholtz Association, Berlin, Germany.,Neuroscience Research Center and Cluster of Excellence NeuroCure, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Carmen Birchmeier
- Developmental Biology/Signal Transduction Group, Max-Delbrueck-Centrum in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
17
|
Neuregulin-2 ablation results in dopamine dysregulation and severe behavioral phenotypes relevant to psychiatric disorders. Mol Psychiatry 2018; 23:1233-1243. [PMID: 28322273 PMCID: PMC5608621 DOI: 10.1038/mp.2017.22] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 11/16/2016] [Accepted: 12/21/2016] [Indexed: 12/17/2022]
Abstract
Numerous genetic and functional studies implicate variants of Neuregulin-1 (NRG1) and its neuronal receptor ErbB4 in schizophrenia and many of its endophenotypes. Although the neurophysiological and behavioral phenotypes of NRG1 mutant mice have been investigated extensively, practically nothing is known about the function of NRG2, the closest NRG1 homolog. We found that NRG2 expression in the adult rodent brain does not overlap with NRG1 and is more extensive than originally reported, including expression in the striatum and medial prefrontal cortex (mPFC), and therefore generated NRG2 knockout mice (KO) to study its function. NRG2 KOs have higher extracellular dopamine levels in the dorsal striatum but lower levels in the mPFC; a pattern with similarities to dopamine dysbalance in schizophrenia. Like ErbB4 KO mice, NRG2 KOs performed abnormally in a battery of behavioral tasks relevant to psychiatric disorders. NRG2 KOs exhibit hyperactivity in a novelty-induced open field, deficits in prepulse inhibition, hypersensitivity to amphetamine, antisocial behaviors, reduced anxiety-like behavior in the elevated plus maze and deficits in the T-maze alteration reward test-a task dependent on hippocampal and mPFC function. Acute administration of clozapine rapidly increased extracellular dopamine levels in the mPFC and improved alternation T-maze performance. Similar to mice treated chronically with N-methyl-d-aspartate receptor (NMDAR) antagonists, we demonstrate that NMDAR synaptic currents in NRG2 KOs are augmented at hippocampal glutamatergic synapses and are more sensitive to ifenprodil, indicating an increased contribution of GluN2B-containing NMDARs. Our findings reveal a novel role for NRG2 in the modulation of behaviors with relevance to psychiatric disorders.
Collapse
|
18
|
Skirzewski M, Karavanova I, Shamir A, Erben L, Garcia-Olivares J, Shin JH, Vullhorst D, Alvarez VA, Amara SG, Buonanno A. ErbB4 signaling in dopaminergic axonal projections increases extracellular dopamine levels and regulates spatial/working memory behaviors. Mol Psychiatry 2018; 23:2227-2237. [PMID: 28727685 PMCID: PMC5775946 DOI: 10.1038/mp.2017.132] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 03/13/2017] [Accepted: 04/04/2017] [Indexed: 02/07/2023]
Abstract
Genetic variants of Neuregulin 1 (NRG1) and its neuronal tyrosine kinase receptor ErbB4 are associated with risk for schizophrenia, a neurodevelopmental disorder characterized by excitatory/inhibitory imbalance and dopamine (DA) dysfunction. To date, most ErbB4 studies have focused on GABAergic interneurons in the hippocampus and neocortex, particularly fast-spiking parvalbumin-positive (PV+) basket cells. However, NRG has also been shown to modulate DA levels, suggesting a role for ErbB4 signaling in dopaminergic neuron function. Here we report that ErbB4 in midbrain DAergic axonal projections regulates extracellular DA levels and relevant behaviors. Mice lacking ErbB4 in tyrosine hydroxylase-positive (TH+) neurons, but not in PV+ GABAergic interneurons, exhibit different regional imbalances of basal DA levels and fail to increase DA in response to local NRG1 infusion into the dorsal hippocampus, medial prefrontal cortex and dorsal striatum measured by reverse microdialysis. Using Lund Human Mesencephalic (LUHMES) cells, we show that NRG/ErbB signaling increases extracellular DA levels, at least in part, by reducing DA transporter (DAT)-dependent uptake. Interestingly, TH-Cre;ErbB4f/f mice manifest deficits in learning, spatial and working memory-related behaviors, but not in numerous other behaviors altered in PV-Cre;ErbB4f/f mice. Importantly, microinjection of a Cre-inducible ErbB4 virus (AAV-ErbB4.DIO) into the mesencephalon of TH-Cre;ErbB4f/f mice, which selectively restores ErbB4 expression in DAergic neurons, rescues DA dysfunction and ameliorates behavioral deficits. Our results indicate that direct NRG/ErbB4 signaling in DAergic axonal projections modulates DA homeostasis, and that NRG/ErbB4 signaling in both GABAergic interneurons and DA neurons contribute to the modulation of behaviors relevant to psychiatric disorders.
Collapse
Affiliation(s)
- M Skirzewski
- 0000 0001 2297 5165grid.94365.3dSection on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - I Karavanova
- 0000 0001 2297 5165grid.94365.3dSection on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - A Shamir
- 0000 0001 2297 5165grid.94365.3dSection on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - L Erben
- 0000 0001 2297 5165grid.94365.3dSection on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA ,0000 0001 2240 3300grid.10388.32Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
| | - J Garcia-Olivares
- 0000 0001 2297 5165grid.94365.3dLaboratory of Molecular and Cellular Neurobiology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD USA
| | - J H Shin
- 0000 0001 2297 5165grid.94365.3dLaboratory for Integrative Neuroscience, Section on Neuronal Structure, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD USA
| | - D Vullhorst
- 0000 0001 2297 5165grid.94365.3dSection on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - V A Alvarez
- 0000 0001 2297 5165grid.94365.3dLaboratory for Integrative Neuroscience, Section on Neuronal Structure, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD USA
| | - S G Amara
- 0000 0001 2297 5165grid.94365.3dLaboratory of Molecular and Cellular Neurobiology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD USA
| | - A Buonanno
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
19
|
|
20
|
Neuregulin-Dependent Regulation of Fast-Spiking Interneuron Excitability Controls the Timing of the Critical Period. J Neurosci 2017; 36:10285-10295. [PMID: 27707966 DOI: 10.1523/jneurosci.4242-15.2016] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 08/01/2016] [Indexed: 02/06/2023] Open
Abstract
Maturation of excitatory drive onto fast-spiking interneurons (FS INs) in the visual cortex has been implicated in the control of the timing of the critical period for ocular dominance plasticity. However, the mechanisms that regulate the strength of these synapses over cortical development are not understood. Here we use a mouse model to show that neuregulin (NRG) and the receptor tyrosine kinase erbB4 regulate the timing of the critical period. NRG1 enhanced the strength of excitatory synapses onto FS INs, which inhibited ocular dominance plasticity during the critical period but rescued plasticity in transgenics with hypoexcitable FS INs. Blocking the effects of endogenous neuregulin via inhibition of erbBs rescued ocular dominance plasticity in postcritical period adults, allowing recovery from amblyopia induced by chronic monocular deprivation. Thus, the strength of excitation onto FS INs is a key determinant of critical period plasticity and is maintained at high levels by NRG-erbB4 signaling to constrain plasticity in adulthood. SIGNIFICANCE STATEMENT Despite decades of experimentation, the mechanisms by which critical periods of enhanced synaptic plasticity are initiated and terminated are not completely understood. Here we show that neuregulin (NRG) and the receptor tyrosine kinase erbB4 determine critical period timing by controlling the strength of excitatory synapses onto FS INs. NRG1 enhanced excitatory drive onto fast spiking interneurons, which inhibited ocular dominance plasticity in juveniles but rescued plasticity in transgenics with hypoexcitable FS INs. Blocking the effects of endogenous neuregulin via inhibition of erbBs rescued ocular dominance plasticity in adults, allowing recovery from amblyopia induced by chronic monocular deprivation. Thus, in contrast to prevailing views of the termination of the critical period, active maintenance of strong excitation onto FS INs constrains plasticity in adults.
Collapse
|
21
|
Erbb4 Deletion from Medium Spiny Neurons of the Nucleus Accumbens Core Induces Schizophrenia-Like Behaviors via Elevated GABA A Receptor α1 Subunit Expression. J Neurosci 2017; 37:7450-7464. [PMID: 28667174 DOI: 10.1523/jneurosci.3948-16.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 06/03/2017] [Accepted: 06/19/2017] [Indexed: 12/21/2022] Open
Abstract
Medium spiny neurons (MSNs), the major GABAergic projection neurons in the striatum, are implicated in many neuropsychiatric diseases such as schizophrenia, but the underlying mechanisms remain unclear. We found that a deficiency in Erbb4, a schizophrenia risk gene, in MSNs of the nucleus accumbens (NAc) core, but not the dorsomedial striatum, markedly induced schizophrenia-like behaviors such as hyperactivity, abnormal marble-burying behavior, damaged social novelty recognition, and impaired sensorimotor gating function in male mice. Using immunohistochemistry, Western blot, RNA interference, electrophysiology, and behavior test studies, we found that these phenomena were mediated by increased GABAA receptor α1 subunit (GABAAR α1) expression, which enhanced inhibitory synaptic transmission on MSNs. These results suggest that Erbb4 in MSNs of the NAc core may contribute to the pathogenesis of schizophrenia by regulating GABAergic transmission and raise the possibility that GABAAR α1 may therefore serve as a new therapeutic target for schizophrenia.SIGNIFICANCE STATEMENT Although ErbB4 is highly expressed in striatal medium spiny neurons (MSNs), its role in this type of neuron has not been reported previously. The present study demonstrates that Erbb4 deletion in nucleus accumbens (NAc) core MSNs can induce schizophrenia-like behaviors via elevated GABAA receptor α1 subunit (GABAAR α1) expression. To our knowledge, this is the first evidence that ErbB4 signaling in the MSNs is involved in the pathology of schizophrenia. Furthermore, restoration of GABAAR α1 in the NAc core, but not the dorsal medium striatum, alleviated the abnormal behaviors. Here, we highlight the role of the NAc core in the pathogenesis of schizophrenia and suggest that GABAAR α1 may be a potential pharmacological target for its treatment.
Collapse
|
22
|
Chung DW, Volk DW, Arion D, Zhang Y, Sampson AR, Lewis DA. Dysregulated ErbB4 Splicing in Schizophrenia: Selective Effects on Parvalbumin Expression. Am J Psychiatry 2016; 173:60-8. [PMID: 26337038 PMCID: PMC5242561 DOI: 10.1176/appi.ajp.2015.15020150] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Alternative splicing of ErbB4 transcripts is dysregulated in the dorsolateral prefrontal cortex in schizophrenia. ErbB4 regulates the activity of parvalbumin interneurons, and therefore dysregulated ErbB4 splicing could contribute to lower parvalbumin interneuron activity and consequently lower parvalbumin levels in schizophrenia. However, ErbB4 is also present in calretinin interneurons, which are not affected in schizophrenia. Therefore, the authors hypothesized that dysregulated ErbB4 splicing occurs selectively in parvalbumin interneurons and is associated with lower parvalbumin levels in schizophrenia. METHOD Tissue samples enriched in calretinin and parvalbumin interneurons were laser microdissected from dorsolateral prefrontal cortex layers 2 and 4, respectively, from matched pairs of schizophrenia and comparison subjects. Transcript levels for pan-ErbB4, four ErbB4 splicing variants (JM-a, JM-b, CYT-1, CYT-2), parvalbumin, and calretinin were quantified by quantitative polymerase chain reaction (qPCR) in each layer. Transcript levels for myocardial infarction associated transcript (MIAT), which regulates ErbB4 splicing, were quantified in gray matter by qPCR and in parvalbumin interneurons by microarray. RESULTS Calretinin and parvalbumin mRNAs were preferentially expressed in layers 2 and 4, respectively. In schizophrenia subjects, lower parvalbumin levels, higher CYT-1 and JM-a levels, and lower CYT-2 and JM-b levels were detected selectively in layer 4. In layer 4, the JM-a/JM-b ratio was inversely correlated with parvalbumin levels in schizophrenia subjects. MIAT levels were preferentially higher in parvalbumin interneurons in schizophrenia subjects. CONCLUSIONS These findings suggest that elevated MIAT expression alters ErbB4 splicing selectively in parvalbumin interneurons in schizophrenia. Dysregulated ErbB4 splicing in schizophrenia may contribute to lower activity of parvalbumin interneurons and an activity-dependent down-regulation of parvalbumin expression.
Collapse
Affiliation(s)
- Daniel W Chung
- From the Translational Neuroscience Program, Department of Psychiatry, and the Medical Scientist Training Program, University of Pittsburgh School of Medicine; and the Department of Statistics, University of Pittsburgh
| | - David W Volk
- From the Translational Neuroscience Program, Department of Psychiatry, and the Medical Scientist Training Program, University of Pittsburgh School of Medicine; and the Department of Statistics, University of Pittsburgh
| | - Dominique Arion
- From the Translational Neuroscience Program, Department of Psychiatry, and the Medical Scientist Training Program, University of Pittsburgh School of Medicine; and the Department of Statistics, University of Pittsburgh
| | - Yun Zhang
- From the Translational Neuroscience Program, Department of Psychiatry, and the Medical Scientist Training Program, University of Pittsburgh School of Medicine; and the Department of Statistics, University of Pittsburgh
| | - Allan R Sampson
- From the Translational Neuroscience Program, Department of Psychiatry, and the Medical Scientist Training Program, University of Pittsburgh School of Medicine; and the Department of Statistics, University of Pittsburgh
| | - David A Lewis
- From the Translational Neuroscience Program, Department of Psychiatry, and the Medical Scientist Training Program, University of Pittsburgh School of Medicine; and the Department of Statistics, University of Pittsburgh
| |
Collapse
|
23
|
Vullhorst D, Mitchell RM, Keating C, Roychowdhury S, Karavanova I, Tao-Cheng JH, Buonanno A. A negative feedback loop controls NMDA receptor function in cortical interneurons via neuregulin 2/ErbB4 signalling. Nat Commun 2015; 6:7222. [PMID: 26027736 PMCID: PMC4451617 DOI: 10.1038/ncomms8222] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 04/17/2015] [Indexed: 12/31/2022] Open
Abstract
The neuregulin receptor ErbB4 is an important modulator of GABAergic interneurons and neural network synchronization. However, little is known about the endogenous ligands that engage ErbB4, the neural processes that activate them or their direct downstream targets. Here we demonstrate, in cultured neurons and in acute slices, that the NMDA receptor is both effector and target of neuregulin 2 (NRG2)/ErbB4 signalling in cortical interneurons. Interneurons co-express ErbB4 and NRG2, and pro-NRG2 accumulates on cell bodies atop subsurface cisternae. NMDA receptor activation rapidly triggers shedding of the signalling-competent NRG2 extracellular domain. In turn, NRG2 promotes ErbB4 association with GluN2B-containing NMDA receptors, followed by rapid internalization of surface receptors and potent downregulation of NMDA but not AMPA receptor currents. These effects occur selectively in ErbB4-positive interneurons and not in ErbB4-negative pyramidal neurons. Our findings reveal an intimate reciprocal relationship between ErbB4 and NMDA receptors with possible implications for the modulation of cortical microcircuits associated with cognitive deficits in psychiatric disorders.
Collapse
Affiliation(s)
- Detlef Vullhorst
- Section on Molecular Neurobiology, Eunice Shriver Kennedy National Institute of Child Health and Human Development, Bethesda, Maryland 20892, USA
| | - Robert M Mitchell
- Section on Molecular Neurobiology, Eunice Shriver Kennedy National Institute of Child Health and Human Development, Bethesda, Maryland 20892, USA
| | - Carolyn Keating
- Section on Molecular Neurobiology, Eunice Shriver Kennedy National Institute of Child Health and Human Development, Bethesda, Maryland 20892, USA
| | - Swagata Roychowdhury
- Section on Molecular Neurobiology, Eunice Shriver Kennedy National Institute of Child Health and Human Development, Bethesda, Maryland 20892, USA
| | - Irina Karavanova
- Section on Molecular Neurobiology, Eunice Shriver Kennedy National Institute of Child Health and Human Development, Bethesda, Maryland 20892, USA
| | - Jung-Hwa Tao-Cheng
- EM Facility, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland 20892, USA
| | - Andres Buonanno
- Section on Molecular Neurobiology, Eunice Shriver Kennedy National Institute of Child Health and Human Development, Bethesda, Maryland 20892, USA
| |
Collapse
|
24
|
Morishita H, Kundakovic M, Bicks L, Mitchell A, Akbarian S. Interneuron epigenomes during the critical period of cortical plasticity: Implications for schizophrenia. Neurobiol Learn Mem 2015; 124:104-10. [PMID: 25849095 DOI: 10.1016/j.nlm.2015.03.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 03/23/2015] [Accepted: 03/25/2015] [Indexed: 10/23/2022]
Abstract
Schizophrenia, a major psychiatric disorder defined by delusions and hallucinations, among other symptoms, often with onset in early adulthood, is potentially associated with molecular and cellular alterations in parvalbumin-expressing fast spiking interneurons and other constituents of the cortical inhibitory GABAergic circuitry. The underlying mechanisms, including the role of disease-associated risk factors operating in adolescence such as drug abuse and social stressors, remain incompletely understood. Here, we summarize emerging findings from animal models, highlighting the ability of parvalbuminergic interneurons (PVI) to induce, during the juvenile period, long-term plastic changes in prefrontal and visual cortex, thereby altering perception, cognition and behavior in the adult. Of note, molecular alterations in PVI from subjects with schizophrenia, including downregulated expression of a subset of GABAergic genes, have also been found in juvenile stress models of the disorder. Some of the transcriptional alterations observed in schizophrenia postmortem brain could be linked to changes in the epigenetic architecture of GABAergic gene promoters, including dysregulated DNA methylation, histone modification patterns and disruption of promoter-enhancer interactions at site of chromosomal loop formations. Therefore, we predict that, in the not-to-distant future, PVI- and other cell-type specific epigenomic mappings in the animal model and human brain will provide novel insights into the pathophysiology of schizophrenia and related psychotic diseases, including the role of cortical GABAergic circuitry in shaping long-term plasticity and cognitive function of the cerebral cortex.
Collapse
Affiliation(s)
- Hirofumi Morishita
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States; Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, United States; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, United States.
| | - Marija Kundakovic
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, United States
| | - Lucy Bicks
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, United States
| | - Amanda Mitchell
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, United States
| | - Schahram Akbarian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, United States.
| |
Collapse
|
25
|
Engel M, Snikeris P, Jenner A, Karl T, Huang XF, Frank E. Neuregulin 1 Prevents Phencyclidine-Induced Behavioral Impairments and Disruptions to GABAergic Signaling in Mice. Int J Neuropsychopharmacol 2015; 18:pyu114. [PMID: 26478928 PMCID: PMC4540095 DOI: 10.1093/ijnp/pyu114] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Substantial evidence from human post-mortem and genetic studies has linked the neurotrophic factor neuregulin 1 (NRG1) to the pathophysiology of schizophrenia. Genetic animal models and in vitro experiments have suggested that altered NRG1 signaling, rather than protein changes, contributes to the symptomatology of schizophrenia. However, little is known about the effect of NRG1 on schizophrenia-relevant behavior and neurotransmission (particularly GABAergic and glutamatergic) in adult animals. METHOD To address this question, we treated adult mice with the extracellular signaling domain of NRG1 and assessed spontaneous locomotor activity and acoustic startle response, as well as extracellular GABA, glutamate, and glycine levels in the prefrontal cortex and hippocampus via microdialysis. Furthermore, we asked whether the effect of NRG1 would differ under schizophrenia-relevant impairments in mice and therefore co-treated mice with NRG1 and phencyclidine (PCP) (3 mg/kg). RESULTS Acute intraventricularly- or systemically-injected NRG1 did not affect spontaneous behavior, but prevented PCP induced hyperlocomotion and deficits of prepulse inhibition. NRG1 retrodialysis (10 nM) reduced extracellular glutamate and glycine levels in the prefrontal cortex and hippocampus, and prevented PCP-induced increase in extracellular GABA levels in the hippocampus. CONCLUSION With these results, we provide the first compelling in vivo evidence for the involvement of NRG1 signaling in schizophrenia-relevant behavior and neurotransmission in the adult nervous system, which highlight its treatment potential. Furthermore, the ability of NRG1 treatment to alter GABA, glutamate, and glycine levels in the presence of PCP also suggests that NRG1 signaling has the potential to alter disrupted neurotransmission in patients with schizophrenia.
Collapse
|
26
|
Schmidt MJ, Mirnics K. Neurodevelopment, GABA system dysfunction, and schizophrenia. Neuropsychopharmacology 2015; 40:190-206. [PMID: 24759129 PMCID: PMC4262918 DOI: 10.1038/npp.2014.95] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/03/2014] [Accepted: 04/11/2014] [Indexed: 02/07/2023]
Abstract
The origins of schizophrenia have eluded clinicians and researchers since Kraepelin and Bleuler began documenting their findings. However, large clinical research efforts in recent decades have identified numerous genetic and environmental risk factors for schizophrenia. The combined data strongly support the neurodevelopmental hypothesis of schizophrenia and underscore the importance of the common converging effects of diverse insults. In this review, we discuss the evidence that genetic and environmental risk factors that predispose to schizophrenia disrupt the development and normal functioning of the GABAergic system.
Collapse
Affiliation(s)
- Martin J Schmidt
- Department of Psychiatry, Vanderbilt University, Nashville, TN, USA
| | - Karoly Mirnics
- Department of Psychiatry, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN, USA
- Department of Psychiatry, University of Szeged, Szeged, Hungary
| |
Collapse
|
27
|
Mei L, Nave KA. Neuregulin-ERBB signaling in the nervous system and neuropsychiatric diseases. Neuron 2014; 83:27-49. [PMID: 24991953 DOI: 10.1016/j.neuron.2014.06.007] [Citation(s) in RCA: 436] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neuregulins (NRGs) comprise a large family of growth factors that stimulate ERBB receptor tyrosine kinases. NRGs and their receptors, ERBBs, have been identified as susceptibility genes for diseases such as schizophrenia (SZ) and bipolar disorder. Recent studies have revealed complex Nrg/Erbb signaling networks that regulate the assembly of neural circuitry, myelination, neurotransmission, and synaptic plasticity. Evidence indicates there is an optimal level of NRG/ERBB signaling in the brain and deviation from it impairs brain functions. NRGs/ERBBs and downstream signaling pathways may provide therapeutic targets for specific neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Lin Mei
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA; Department of Neurology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA; Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Goettingen, Germany.
| |
Collapse
|